1
|
Zhang D, Kukkar D, Bhatt P, Kim KH, Kaur K, Wang J. Novel nanomaterials-based combating strategies against drug-resistant bacteria. Colloids Surf B Biointerfaces 2025; 248:114478. [PMID: 39778220 DOI: 10.1016/j.colsurfb.2024.114478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Numerous types of contemporary antibiotic treatment regimens have become ineffective with the increasing incidence of drug tolerance. As a result, it is pertinent to seek novel and innovative solutions such as antibacterial nanomaterials (NMs) for the prohibition and treatment of hazardous microbial infections. Unlike traditional antibiotics (e.g., penicillin and tetracycline), the unique physicochemical characteristics (e.g., size dependency) of NMs endow them with bacteriostatic and bactericidal potential. However, it is yet difficult to mechanistically predict or decipher the networks of molecular interaction (e.g., between NMs and the biological systems) and the subsequent immune responses. In light of such research gap, this review outlines various mechanisms accountable for the inception of drug tolerance in bacteria. It also delineates the primary factors governing the NMs-induced molecular mechanisms against microbes, specifically drug-resistant bacteria along with the various NM-based mechanisms of antibacterial activity. The review also explores future directions and prospects for NMs in combating drug-resistant bacteria, while addressing challenges to their commercial viability within the healthcare industry.
Collapse
Affiliation(s)
- Daohong Zhang
- Yantai Key Laboratory of Nanoscience and Technology for Prepared Food, Yantai Engineering Research Center of Green Food Processing and Quality Control, College of Food Engineering, Ludong University, Yantai, Shandong 264025, China
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, South Korea.
| | - Kamalpreet Kaur
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab 140406, India
| | - Jianlong Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
2
|
Yang K, Zhao J, Wang T, Wang Z, Sun R, Gu D, Liu H, Wang W, Zhang C, Zhao C, Guo Y, Ma J, Wei B. Clinical application of targeted next-generation sequencing in pneumonia diagnosis among cancer patients. Front Cell Infect Microbiol 2025; 15:1497198. [PMID: 40041142 PMCID: PMC11876428 DOI: 10.3389/fcimb.2025.1497198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Background Cancer patients are highly susceptible to infections due to their immunocompromised state from both the malignancy and intensive treatments. Accurate and timely identification of causative pathogens is crucial for effective management and treatment. Targeted next-generation sequencing (tNGS) has become an important tool in clinical infectious disease diagnosis because of its broad microbial detection range and acceptable cost. However, there is currently a lack of systematic research to evaluate the diagnostic value of this method in cancer patients. Methods To evaluate the diagnostic value of tNGS for cancer patients with pneumonia, a retrospective analysis was conducted on 148 patients with suspected pneumonia who were treated at the Henan Cancer Hospital. The tNGS results were compared with conventional microbiological tests (CMT) and clinical diagnoses based on symptoms and imaging studies to assess the diagnostic performance of tNGS in cancer patients with pneumonia. Results Among these 148 patients, 130 were ultimately diagnosed with pneumonia. tNGS demonstrated significantly higher sensitivity (84.62% vs. 56.92%) and diagnostic accuracy (85.81% vs. 62.16%) compared to the CMT method. The tNGS method identified more pathogens than CMT method (87.50% vs 57.14%), regardless of whether they were bacteria, fungi, or viruses, primarily due to its broader pathogen detection range and higher sensitivity compared to the CMT method. tNGS had significantly higher diagnostic accuracy for Pneumocystis jirovecii and Legionella pneumophila than the CMT method, but for most pathogens, tNGS showed higher sensitivity but with a correspondingly lower specificity compared to CMT. Conclusion tNGS demonstrates higher sensitivity and a broader pathogen detection spectrum compared to CMT, making it a valuable diagnostic tool for managing pneumonia in cancer patients.
Collapse
Affiliation(s)
- Ke Yang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jiuzhou Zhao
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Tingjie Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zhizhong Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Rui Sun
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Dejian Gu
- Medical Department, Geneplus-Beijing Co., Ltd., Beijing, China
| | - Hao Liu
- Medical Department, Geneplus-Beijing Co., Ltd., Beijing, China
| | - Weizhen Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Cuiyun Zhang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Chengzhi Zhao
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yongjun Guo
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jie Ma
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Bing Wei
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
3
|
Stévenin V, Coipan CE, Duijster JW, van Elsland DM, Voogd L, Bigey L, van Hoek AHAM, Wijnands LM, Janssen L, Akkermans JJLL, Neefjes-Borst A, Franz E, Mughini-Gras L, Neefjes J. Multi-omics analyses of cancer-linked clinical salmonellae reveal bacterial-induced host metabolic shift and mTOR-dependent cell transformation. Cell Rep 2024; 43:114931. [PMID: 39488829 DOI: 10.1016/j.celrep.2024.114931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
Salmonellae are associated epidemiologically and experimentally with colon cancer. To understand how Salmonella induces cell transformation, we performed multi-omics and phenotypic analyses of Salmonella clinical strains isolated from patients later diagnosed with colon cancer (case strains) and control strains from patients without cancer. We show that high transformation efficiency is a frequent intrinsic feature of clinical (case and control) salmonellae, yet case strains showed higher transformation efficiency than control strains. Transformation efficiency correlates with gene expression, nutrient utilization, and intracellular virulence, but not with genetic features, suggesting a phenotypic convergence of Salmonella strains resulting in cell transformation. We show that both bacterial entry and intracellular replication are required for host cell transformation and are associated with hyperactivation of the mTOR pathway. Strikingly, transiently inactivating mTOR through chemical inhibition reverses the transformation phenotype instigated by Salmonella infection. This suggests that targeting the mTOR pathway could prevent the development of Salmonella-induced tumors.
Collapse
Affiliation(s)
- Virginie Stévenin
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), 2333 ZC Leiden, the Netherlands.
| | - Claudia E Coipan
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Janneke W Duijster
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Daphne M van Elsland
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), 2333 ZC Leiden, the Netherlands
| | - Linda Voogd
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), 2333 ZC Leiden, the Netherlands
| | - Lise Bigey
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), 2333 ZC Leiden, the Netherlands; École Normale Supérieure Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Angela H A M van Hoek
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Lucas M Wijnands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Lennert Janssen
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), 2333 ZC Leiden, the Netherlands
| | - Jimmy J L L Akkermans
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), 2333 ZC Leiden, the Netherlands
| | - Andra Neefjes-Borst
- Pathology Department, Amsterdam University Medical Center (VUmc), 1081 HV Amsterdam, the Netherlands
| | - Eelco Franz
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Lapo Mughini-Gras
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands; Institute for Risk Assessment Sciences, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), 2333 ZC Leiden, the Netherlands.
| |
Collapse
|
4
|
Yao R, Sun L, Gao R, Mei Y, Xue G, Yu D. PTTM: dissecting the profile of tumor tissue microbiome to reveal microbiota features and associations with host transcriptome. Brief Bioinform 2024; 26:bbaf057. [PMID: 39924716 PMCID: PMC11807729 DOI: 10.1093/bib/bbaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/05/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025] Open
Abstract
Microbiota is present in the human tissue microenvironment and closely related to tumorigenesis and treatment. However, the landscape of tissue microbiome and its relationship with tumors remain less understood. In this study, we re-analyzed the omics data from the 7104 samples (94 projects for 15 cancers) in the NCBI database to obtain microbial profiles. After normalization and decontamination processing, we established classification models to distinguish between different tumors and tumor with adjacent normal tissues. The models had excellent performances, indicating that tissue microbiome had significant tumor specificity. Moreover, a series of key bacteria and bacteria-gene association pairs were screened out based on bioinformatic analysis, such as the tumor-promoting bacteria Fusobacterium, the tumor-suppressing bacteria Actinomyces, and the significant Rhodopseudomonas-COL1A1 association pair. In addition, we created a visual website, PTTM (http://198.46.152.196:7080/), for users to query and download the results. The identified key bacteria and association pairs provide candidate targets for further exploration of the molecular mechanisms of microbial action on tumorigenesis and the development of cancer therapy.
Collapse
Affiliation(s)
- Ruiqian Yao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- Department of Medical Genetics, Naval Medical University, Xiang-Yin Road, 800, Shanghai 200433, China
- Department of Dermatology, Naval Medical Centre, Naval Medical University, Shanghai 200052, China
| | - Lu Sun
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Xiang-Yin Road, 800, Shanghai 200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| | - Ruifang Gao
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Xiang-Yin Road, 800, Shanghai 200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| | - Yue Mei
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Xiang-Yin Road, 800, Shanghai 200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| | - Geng Xue
- Department of Medical Genetics, Naval Medical University, Xiang-Yin Road, 800, Shanghai 200433, China
| | - Dong Yu
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Xiang-Yin Road, 800, Shanghai 200433, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| |
Collapse
|
5
|
Yan B, Zeng L, Lu Y, Li M, Lu W, Zhou B, He Q. Rapid bacterial identification through volatile organic compound analysis and deep learning. BMC Bioinformatics 2024; 25:347. [PMID: 39506632 PMCID: PMC11539783 DOI: 10.1186/s12859-024-05967-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND The increasing antimicrobial resistance caused by the improper use of antibiotics poses a significant challenge to humanity. Rapid and accurate identification of microbial species in clinical settings is crucial for precise medication and reducing the development of antimicrobial resistance. This study aimed to explore a method for automatic identification of bacteria using Volatile Organic Compounds (VOCs) analysis and deep learning algorithms. RESULTS AlexNet, where augmentation is applied, produces the best results. The average accuracy rate for single bacterial culture classification reached 99.24% using cross-validation, and the accuracy rates for identifying the three bacteria in randomly mixed cultures were SA:98.6%, EC:98.58% and PA:98.99%, respectively. CONCLUSION This work provides a new approach to quickly identify bacterial microorganisms. Using this method can automatically identify bacteria in GC-IMS detection results, helping clinical doctors quickly detect bacterial species, accurately prescribe medication, thereby controlling epidemics, and minimizing the negative impact of bacterial resistance on society.
Collapse
Affiliation(s)
- Bowen Yan
- Research Department, Daping Hosipital, Army Medical University, Chongqing, 400042, China
| | - Lin Zeng
- Research Department, Daping Hosipital, Army Medical University, Chongqing, 400042, China
| | - Yanyi Lu
- Research Department, Daping Hosipital, Army Medical University, Chongqing, 400042, China
| | - Min Li
- Laboratory Department, Daping Hosipital, Army Medical University, Chongqing, 400042, China
| | - Weiping Lu
- Laboratory Department, Daping Hosipital, Army Medical University, Chongqing, 400042, China
| | - Bangfu Zhou
- Research Department, Daping Hosipital, Army Medical University, Chongqing, 400042, China
| | - Qinghua He
- Research Department, Daping Hosipital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
6
|
Halawa M, Newman PM, Aderibigbe T, Carabetta VJ. Conjugated therapeutic proteins as a treatment for bacteria which trigger cancer development. iScience 2024; 27:111029. [PMID: 39635133 PMCID: PMC11615139 DOI: 10.1016/j.isci.2024.111029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
In recent years, an increasing amount of research has focused on the intricate and complex correlation between bacterial infections and the development of cancer. Some studies even identified specific bacterial species as potential culprits in the initiation of carcinogenesis, which generated a great deal of interest in the creation of innovative therapeutic strategies aimed at addressing both the infection and the subsequent risk of cancer. Among these strategies, there has been a recent emergence of the use of conjugated therapeutic proteins, which represent a highly promising avenue in the field of cancer therapeutics. These proteins offer a dual-targeting approach that seeks to effectively combat both the bacterial infection and the resulting malignancies that may arise because of such infections. This review delves into the landscape of conjugated therapeutic proteins that have been intricately designed with the purpose of specifically targeting bacteria that have been implicated in the induction of cancer.
Collapse
Affiliation(s)
- Mohamed Halawa
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Precious M. Newman
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Tope Aderibigbe
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Valerie J. Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
7
|
Mikó E, Sipos A, Tóth E, Lehoczki A, Fekete M, Sebő É, Kardos G, Bai P. Guideline for designing microbiome studies in neoplastic diseases. GeroScience 2024; 46:4037-4057. [PMID: 38922379 PMCID: PMC11336004 DOI: 10.1007/s11357-024-01255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Oncobiosis has emerged as a key contributor to the development, and modulator of the treatment efficacy of cancer. Hereby, we review the modalities through which the oncobiome can support the progression of tumors, and the emerging therapeutic opportunities they present. The review highlights the inherent challenges and limitations faced in sampling and accurately characterizing oncobiome. Additionally, the review underscores the critical need for the standardization of microbial analysis techniques and the consistent reporting of microbiome data. We provide a suggested metadata set that should accompany microbiome datasets from oncological settings so that studies remain comparable and decipherable.
Collapse
Affiliation(s)
- Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary
| | - Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group, 4032, Debrecen, Hungary
| | - Andrea Lehoczki
- Department of Hematology and Stem Cell Transplantation, South Pest Central Hospital-National Institute for Hematology and Infectious Diseases, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Éva Sebő
- Breast Center, Kenézy Gyula Hospital, University of Debrecen, 4032, Debrecen, Hungary
| | - Gábor Kardos
- Department of Metagenomics, University of Debrecen, 4032, Debrecen, Hungary
- Faculty of Health Sciences, One Health Institute, University of Debrecen, 4032, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1., 4032, Debrecen, Hungary.
- HUN-REN-DE Cell Biology and Signaling Research Group, 4032, Debrecen, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032, Debrecen, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
- Center of Excellence, The Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
8
|
Yang X, Gan Y, Zhang Y, Liu Z, Geng J, Wang W. Microbial genotoxin-elicited host DNA mutations related to mitochondrial dysfunction, a momentous contributor for colorectal carcinogenesis. mSystems 2024; 9:e0088724. [PMID: 39189772 PMCID: PMC11406885 DOI: 10.1128/msystems.00887-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024] Open
Abstract
Gut microbe dysbiosis increases repetitive inflammatory responses, leading to an increase in the incidence of colorectal cancer. Recent studies have revealed that specific microbial species directly instigate mutations in the host nucleus DNA, thereby accelerating the progression of colorectal cancer. Given the well-established role of mitochondrial dysfunction in promoting colorectal cancer, it is reasonable to postulate that gut microbes may induce mitochondrial gene mutations, thereby inducing mitochondrial dysfunction. In this review, we focus on gut microbial genotoxins and their known and potential targets in mitochondrial genes. Consequently, we propose that targeted disruption of genotoxin transport pathways may effectively reduce the rate of mitochondrial gene mutations and yield substantial benefits for the prevention of colorectal carcinogenesis.
Collapse
Affiliation(s)
- Xue Yang
- Department of Infectious Disease and Hepatic Disease, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yumeng Gan
- Department of Infectious Disease and Hepatic Disease, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yuting Zhang
- Department of Infectious Disease and Hepatic Disease, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zhongjian Liu
- Institute of Basic and Clinical Medicine, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiawei Geng
- Department of Infectious Disease and Hepatic Disease, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Wenxue Wang
- Department of Infectious Disease and Hepatic Disease, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
9
|
Chen B, Yang Y, Wang X, Yang W, Lu Y, Wang D, Zhuo E, Tang Y, Su J, Tang G, Shao S, Gu K. mRNA vaccine development and applications: A special focus on tumors (Review). Int J Oncol 2024; 65:81. [PMID: 38994758 PMCID: PMC11251742 DOI: 10.3892/ijo.2024.5669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/20/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer is characterized by unlimited proliferation and metastasis, and traditional therapeutic strategies usually result in the acquisition of drug resistance, thus highlighting the need for more personalized treatment. mRNA vaccines transfer the gene sequences of exogenous target antigens into human cells through transcription and translation to stimulate the body to produce specific immune responses against the encoded proteins, so as to enable the body to obtain immune protection against said antigens; this approach may be adopted for personalized cancer therapy. Since the recent coronavirus pandemic, the development of mRNA vaccines has seen substantial progress and widespread adoption. In the present review, the development of mRNA vaccines, their mechanisms of action, factors influencing their function and the current clinical applications of the vaccine are discussed. A focus is placed on the application of mRNA vaccines in cancer, with the aim of highlighting unique advances and the remaining challenges of this novel and promising therapeutic approach.
Collapse
Affiliation(s)
- Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yipin Yang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xinyi Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Wenzhi Yang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - You Lu
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Daoyue Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Enba Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yanchao Tang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Junhong Su
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Guozheng Tang
- Department of Orthopedics, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui 237008, P.R. China
| | - Song Shao
- Department of Orthopedics, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui 237008, P.R. China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
10
|
Kwon SY, Thi-Thu Ngo H, Son J, Hong Y, Min JJ. Exploiting bacteria for cancer immunotherapy. Nat Rev Clin Oncol 2024; 21:569-589. [PMID: 38840029 DOI: 10.1038/s41571-024-00908-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/07/2024]
Abstract
Immunotherapy has revolutionized the treatment of cancer but continues to be constrained by limited response rates, acquired resistance, toxicities and high costs, which necessitates the development of new, innovative strategies. The discovery of a connection between the human microbiota and cancer dates back 4,000 years, when local infection was observed to result in tumour eradication in some individuals. However, the true oncological relevance of the intratumoural microbiota was not recognized until the turn of the twentieth century. The intratumoural microbiota can have pivotal roles in both the pathogenesis and treatment of cancer. In particular, intratumoural bacteria can either promote or inhibit cancer growth via remodelling of the tumour microenvironment. Over the past two decades, remarkable progress has been made preclinically in engineering bacteria as agents for cancer immunotherapy; some of these bacterial products have successfully reached the clinical stages of development. In this Review, we discuss the characteristics of intratumoural bacteria and their intricate interactions with the tumour microenvironment. We also describe the many strategies used to engineer bacteria for use in the treatment of cancer, summarizing contemporary data from completed and ongoing clinical trials. The work described herein highlights the potential of bacteria to transform the landscape of cancer therapy, bridging ancient wisdom with modern scientific innovation.
Collapse
Affiliation(s)
- Seong-Young Kwon
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, Republic of Korea
| | - Hien Thi-Thu Ngo
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeonnam, Republic of Korea
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
| | - Jinbae Son
- CNCure Biotech, Jeonnam, Republic of Korea
| | - Yeongjin Hong
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea
- CNCure Biotech, Jeonnam, Republic of Korea
- Department of Microbiology and Immunology, Chonnam National University Medical School, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University, Jeonnam, Republic of Korea
| | - Jung-Joon Min
- Institute for Molecular Imaging and Theranostics, Chonnam National University Medical School, Jeonnam, Republic of Korea.
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, Republic of Korea.
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeonnam, Republic of Korea.
- CNCure Biotech, Jeonnam, Republic of Korea.
- Department of Microbiology and Immunology, Chonnam National University Medical School, Jeonnam, Republic of Korea.
- National Immunotherapy Innovation Center, Chonnam National University, Jeonnam, Republic of Korea.
| |
Collapse
|
11
|
Shi L, Han X, Liu F, Long J, Jin Y, Chen S, Duan G, Yang H. Review on Long Non-Coding RNAs as Biomarkers and Potentially Therapeutic Targets for Bacterial Infections. Curr Issues Mol Biol 2024; 46:7558-7576. [PMID: 39057090 PMCID: PMC11276060 DOI: 10.3390/cimb46070449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The confrontation between humans and bacteria is ongoing, with strategies for combating bacterial infections continually evolving. With the advancement of RNA sequencing technology, non-coding RNAs (ncRNAs) associated with bacterial infections have garnered significant attention. Recently, long ncRNAs (lncRNAs) have been identified as regulators of sterile inflammatory responses and cellular defense against live bacterial pathogens. They are involved in regulating host antimicrobial immunity in both the nucleus and cytoplasm. Increasing evidence indicates that lncRNAs are critical for the intricate interactions between host and pathogen during bacterial infections. This paper emphatically elaborates on the potential applications of lncRNAs in clinical hallmarks, cellular damage, immunity, virulence, and drug resistance in bacterial infections in greater detail. Additionally, we discuss the challenges and limitations of studying lncRNAs in the context of bacterial infections and highlight clear directions for this promising field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (L.S.); (X.H.); (F.L.); (J.L.); (Y.J.); (S.C.); (G.D.)
| |
Collapse
|
12
|
He Q, Yuan H, Bu Y, Hu J, Olatunde OZ, Gong L, Wang P, Hu T, Li Y, Lu C. Mesoporous Oxidized Mn-Ca Nanoparticles as Potential Antimicrobial Agents for Wound Healing. Molecules 2024; 29:2960. [PMID: 38998912 PMCID: PMC11243354 DOI: 10.3390/molecules29132960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/07/2024] [Accepted: 06/16/2024] [Indexed: 07/14/2024] Open
Abstract
Managing chronic non-healing wounds presents a significant clinical challenge due to their frequent bacterial infections. Mesoporous silica-based materials possess robust wound-healing capabilities attributed to their renowned antimicrobial properties. The current study details the advancement of mesoporous silicon-loaded MnO and CaO molecules (HMn-Ca) against bacterial infections and chronic non-healing wounds. HMn-Ca was synthesized by reducing manganese chloride and calcium chloride by urotropine solution with mesoporous silicon as the template, thereby transforming the manganese and calcium ions on the framework of mesoporous silicon. The developed HMn-Ca was investigated using scanning electron microscopy (SEM), transmission electron microscope (TEM), ultraviolet-visible (UV-visible), and visible spectrophotometry, followed by the determination of Zeta potential. The production of reactive oxygen species (ROS) was determined by using the 3,3,5,5-tetramethylbenzidine (TMB) oxidation reaction. The wound healing effectiveness of the synthesized HMn-Ca is evaluated in a bacterial-infected mouse model. The loading of MnO and CaO inside mesoporous silicon enhanced the generation of ROS and the capacity of bacterial capture, subsequently decomposing the bacterial membrane, leading to the puncturing of the bacterial membrane, followed by cellular demise. As a result, treatment with HMn-Ca could improve the healing of the bacterial-infected wound, illustrating a straightforward yet potent method for engineering nanozymes tailored for antibacterial therapy.
Collapse
Affiliation(s)
- Qianfeng He
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Hui Yuan
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Youshen Bu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Jiangshan Hu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Olagoke Zacchaeus Olatunde
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Lijie Gong
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Peiyuan Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Ting Hu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yuhang Li
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen 361023, China
| | - Canzhong Lu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (Q.H.); (H.Y.); (Y.B.); (J.H.); (O.Z.O.); (L.G.); (P.W.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| |
Collapse
|
13
|
Bloise N, Giannaccari M, Guagliano G, Peluso E, Restivo E, Strada S, Volpini C, Petrini P, Visai L. Growing Role of 3D In Vitro Cell Cultures in the Study of Cellular and Molecular Mechanisms: Short Focus on Breast Cancer, Endometriosis, Liver and Infectious Diseases. Cells 2024; 13:1054. [PMID: 38920683 PMCID: PMC11201503 DOI: 10.3390/cells13121054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Over the past decade, the development of three-dimensional (3D) models has increased exponentially, facilitating the unravelling of fundamental and essential cellular mechanisms by which cells communicate with each other, assemble into tissues and organs and respond to biochemical and biophysical stimuli under both physiological and pathological conditions. This section presents a concise overview of the most recent updates on the significant contribution of different types of 3D cell cultures including spheroids, organoids and organ-on-chip and bio-printed tissues in advancing our understanding of cellular and molecular mechanisms. The case studies presented include the 3D cultures of breast cancer (BC), endometriosis, the liver microenvironment and infections. In BC, the establishment of 3D culture models has permitted the visualization of the role of cancer-associated fibroblasts in the delivery of exosomes, as well as the significance of the physical properties of the extracellular matrix in promoting cell proliferation and invasion. This approach has also become a valuable tool in gaining insight into general and specific mechanisms of drug resistance. Given the considerable heterogeneity of endometriosis, 3D models offer a more accurate representation of the in vivo microenvironment, thereby facilitating the identification and translation of novel targeted therapeutic strategies. The advantages provided by 3D models of the hepatic environment, in conjunction with the high throughput characterizing various platforms, have enabled the elucidation of complex molecular mechanisms underlying various threatening hepatic diseases. A limited number of 3D models for gut and skin infections have been developed. However, a more profound comprehension of the spatial and temporal interactions between microbes, the host and their environment may facilitate the advancement of in vitro, ex vivo and in vivo disease models. Additionally, it may pave the way for the development of novel therapeutic approaches in diverse research fields. The interested reader will also find concluding remarks on the challenges and prospects of using 3D cell cultures for discovering cellular and molecular mechanisms in the research areas covered in this review.
Collapse
Affiliation(s)
- Nora Bloise
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| | - Marialaura Giannaccari
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Giuseppe Guagliano
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
| | - Emanuela Peluso
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Elisa Restivo
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Silvia Strada
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Cristina Volpini
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Paola Petrini
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of Politecnico di Milano, 20133 Milan, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
14
|
Rodrigues R, Sousa C, Vale N. Deciphering the Puzzle: Literature Insights on Chlamydia trachomatis-Mediated Tumorigenesis, Paving the Way for Future Research. Microorganisms 2024; 12:1126. [PMID: 38930508 PMCID: PMC11205399 DOI: 10.3390/microorganisms12061126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Some infectious agents have the potential to cause specific modifications in the cellular microenvironment that could be propitious to the carcinogenesis process. Currently, there are specific viruses and bacteria, such as human papillomavirus (HPV) and Helicobacter pylori, that are well established as risk factors for neoplasia. Chlamydia trachomatis (CT) infections are one of the most common bacterial sexually transmitted infections worldwide, and recent European data confirmed a continuous rise across Europe. The infection is often asymptomatic in both sexes, requiring a screening program for early detection. Notwithstanding, not all countries in Europe have it. Chlamydia trachomatis can cause chronic and persistent infections, resulting in inflammation, and there are plausible biological mechanisms that link the genital infection with tumorigenesis. Herein, we aimed to understand the epidemiological and biological plausibility of CT genital infections causing endometrial, ovarian, and cervical tumors. Also, we covered some of the best suitable in vitro techniques that could be used to study this potential association. In addition, we defend the point of view of a personalized medicine strategy to treat those patients through the discovery of some biomarkers that could allow it. This review supports the need for the development of further fundamental studies in this area, in order to investigate and establish the role of chlamydial genital infections in oncogenesis.
Collapse
Affiliation(s)
- Rafaela Rodrigues
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (C.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, 4465-671 Leça do Balio, Portugal
| | - Carlos Sousa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (C.S.)
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, 4465-671 Leça do Balio, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal; (R.R.); (C.S.)
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
15
|
Sayed IM, Vo DT, Alcantara J, Inouye KM, Pranadinata RF, Luo L, Boland CR, Goyal NP, Kuo DJ, Huang SC, Sahoo D, Ghosh P, Das S. Molecular Signatures for Microbe-Associated Colorectal Cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.26.595902. [PMID: 38853996 PMCID: PMC11160670 DOI: 10.1101/2024.05.26.595902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Genetic factors and microbial imbalances play crucial roles in colorectal cancers (CRCs), yet the impact of infections on cancer initiation remains poorly understood. While bioinformatic approaches offer valuable insights, the rising incidence of CRCs creates a pressing need to precisely identify early CRC events. We constructed a network model to identify continuum states during CRC initiation spanning normal colonic tissue to pre-cancer lesions (adenomatous polyps) and examined the influence of microbes and host genetics. Methods A Boolean network was built using a publicly available transcriptomic dataset from healthy and adenoma affected patients to identify an invariant Microbe-Associated Colorectal Cancer Signature (MACS). We focused on Fusobacterium nucleatum ( Fn ), a CRC-associated microbe, as a model bacterium. MACS-associated genes and proteins were validated by RT-qPCR, RNA seq, ELISA, IF and IHCs in tissues and colon-derived organoids from genetically predisposed mice ( CPC-APC Min+/- ) and patients (FAP, Lynch Syndrome, PJS, and JPS). Results The MACS that is upregulated in adenomas consists of four core genes/proteins: CLDN2/Claudin-2 (leakiness), LGR5/leucine-rich repeat-containing receptor (stemness), CEMIP/cell migration-inducing and hyaluronan-binding protein (epithelial-mesenchymal transition) and IL8/Interleukin-8 (inflammation). MACS was induced upon Fn infection, but not in response to infection with other enteric bacteria or probiotics. MACS induction upon Fn infection was higher in CPC-APC Min+/- organoids compared to WT controls. The degree of MACS expression in the patient-derived organoids (PDOs) generally corresponded with the known lifetime risk of CRCs. Conclusions Computational prediction followed by validation in the organoid-based disease model identified the early events in CRC initiation. MACS reveals that the CRC-associated microbes induce a greater risk in the genetically predisposed hosts, suggesting its potential use for risk prediction and targeted cancer prevention.
Collapse
|
16
|
Bouyahya A, Bakrim S, Aboulaghras S, El Kadri K, Aanniz T, Khalid A, Abdalla AN, Abdallah AA, Ardianto C, Ming LC, El Omari N. Bioactive compounds from nature: Antioxidants targeting cellular transformation in response to epigenetic perturbations induced by oxidative stress. Biomed Pharmacother 2024; 174:116432. [PMID: 38520868 DOI: 10.1016/j.biopha.2024.116432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
Oxidative stress results from a persistent imbalance in oxidation levels that promotes oxidants, playing a crucial role in the early and sustained phases of DNA damage and genomic and epigenetic instability, both of which are intricately linked to the development of tumors. The molecular pathways contributing to carcinogenesis in this context, particularly those related to double-strand and single-strand breaks in DNA, serve as indicators of DNA damage due to oxidation in cancer cases, as well as factors contributing to epigenetic instability through ectopic expressions. Oxidative stress has been considered a therapeutic target for many years, and an increasing number of studies have highlighted the promising effectiveness of natural products in cancer treatment. In this regard, we present significant research on the therapeutic targeting of oxidative stress using natural molecules and underscore the essential role of oxidative stress in cancer. The consequences of stress, especially epigenetic instability, also offer significant therapeutic prospects. In this context, the use of natural epi-drugs capable of modulating and reorganizing the epigenetic network is beginning to emerge remarkably. In this review, we emphasize the close connections between oxidative stress, epigenetic instability, and tumor transformation, while highlighting the role of natural substances as antioxidants and epi-drugs in the anti-tumoral context.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Sara Aboulaghras
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Tarik Aanniz
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Rabat Medical & Pharmacy School, Mohammed V University in Rabat, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan PO Box: 114, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ahmed A Abdallah
- Department of Anatomy, Faculty of Medicine, Umm Alqura University, Makkah 21955, Saudi Arabia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia.
| | - Long Chiau Ming
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia; School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia; Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam.
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| |
Collapse
|
17
|
Khasawneh AI, Himsawi N, Sammour A, Al Shboul S, Alorjani M, Al-Momani H, Shahin U, Al-Momani H, Alotaibi MR, Saleh T. Association of Human Papilloma Virus, Cytomegalovirus, and Epstein-Barr Virus with Breast Cancer in Jordanian Women. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:699. [PMID: 38792882 PMCID: PMC11122978 DOI: 10.3390/medicina60050699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: The investigation of oncogenic viruses and their potential association with breast cancer (BC) remains an intriguing area of study. The current work aims to assess evidence of three specific viruses, human papillomavirus (HPV), cytomegalovirus (CMV), and Epstein-Barr virus (EBV) in BC samples and to explore their relationship with relevant clinicopathological variables. Materials and Methods: The analysis involved BC samples from 110 Jordanian female patients diagnosed with BC and breast tissue samples from 30 control patients with no evidence of breast malignancy, investigated using real-time PCR. The findings were then correlated with various clinico-pathological characteristics of BC. Results: HPV was detected in 27 (24.5%), CMV in 15 (13.6%), and EBV in 18 (16.4%) BC patients. None of the control samples was positive for HPV or CMV while EBV was detected in only one (3.3%) sample. While (HPV/EBV), (HPV/CMV), and (EBV/CMV) co-infections were reported in 1.8%, 2.7%, and 5.5%, respectively, coinfection with the three viruses (HPV/CMV/EBV) was not reported in our cohort. A statistically significant association was observed between HPV status and age (p = 0.047), and between clinical stage and CMV infection (p = 0.015). Conclusions: Our findings indicate the presence or co-presence of HPV, CMV, and EBV in the BC subpopulation, suggesting a potential role in its development and/or progression. Further investigation is required to elucidate the underlying mechanisms that account for the exact role of oncoviruses in breast carcinogenesis.
Collapse
Affiliation(s)
- Ashraf I. Khasawneh
- Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan (H.A.-M.)
| | - Nisreen Himsawi
- Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan (H.A.-M.)
| | - Ashraf Sammour
- Department of Anatomy, Physiology & Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Sofian Al Shboul
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Mohammed Alorjani
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Hadeel Al-Momani
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Uruk Shahin
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Hafez Al-Momani
- Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan (H.A.-M.)
| | - Moureq R. Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11421, Saudi Arabia
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| |
Collapse
|
18
|
Baldelli G, De Santi M, Ateba CN, Cifola G, Amagliani G, Tchatchouang CDK, Montso PK, Brandi G, Schiavano GF. The potential role of Listeria monocytogenes in promoting colorectal adenocarcinoma tumorigenic process. BMC Microbiol 2024; 24:87. [PMID: 38491424 PMCID: PMC10941472 DOI: 10.1186/s12866-024-03240-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Listeria monocytogenes is a foodborne pathogen, which can cause a severe illness, especially in people with a weakened immune system or comorbidities. The interactions between host and pathogens and between pathogens and tumor cells have been debated in recent years. However, it is still unclear how bacteria can interact with tumor cells, and if this interaction can affect tumor progression and therapy. METHODS In this study, we evaluated the involvement of L. monocytogenes in pre-neoplastic and colorectal cancer cell proliferation and tumorigenic potential. RESULTS Our findings showed that the interaction between heat-killed L. monocytogenes and pre-neoplastic or colorectal cancer cells led to a proliferative induction; furthermore, by using a three-dimensional cell culture model, the obtained data indicated that L. monocytogenes was able to increase the tumorigenic potential of both pre-neoplastic and colorectal cancer cells. The observed effects were then confirmed as L. monocytogenes-specific, using Listeria innocua as negative control. Lastly, data suggested the Insulin Growth Factor 1 Receptor (IGF1R) cascade as one of the possible mechanisms involved in the effects induced by L. monocytogenes in the human colorectal adenocarcinoma cell line. CONCLUSIONS These findings, although preliminary, suggest that the presence of pathogenic bacterial cells in the tumor niches may directly induce, increase, and stimulate tumor progression.
Collapse
Affiliation(s)
- Giulia Baldelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Urbino, Italy
| | - Mauro De Santi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Urbino, Italy
| | - Collins Njie Ateba
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho, South Africa
| | - Giorgia Cifola
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Urbino, Italy
| | - Giulia Amagliani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Urbino, Italy
| | | | - Peter Kotsoana Montso
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho, South Africa
| | - Giorgio Brandi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Urbino, Italy
| | | |
Collapse
|
19
|
Travaglione S, Carlini F, Maroccia Z, Fabbri A. Special Issue "Bacterial Toxins and Cancer". Int J Mol Sci 2024; 25:2128. [PMID: 38396805 PMCID: PMC10889233 DOI: 10.3390/ijms25042128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Infection is a major contributor to the development of cancer, with more than 15% of new cancer diagnoses estimated to be caused by infection [...].
Collapse
Affiliation(s)
| | | | | | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.T.); (F.C.); (Z.M.)
| |
Collapse
|
20
|
Janeczko M, Kochanowicz E, Górka K, Skrzypek T. Quinalizarin as a potential antifungal drug for the treatment of Candida albicans fungal infection in cancer patients. Microbiol Spectr 2024; 12:e0365223. [PMID: 38289929 PMCID: PMC10913734 DOI: 10.1128/spectrum.03652-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024] Open
Abstract
This study aims to analyze the antifungal properties of quinalizarin, a plant-derived compound with proven anticancer effects. Quinalizarin exhibited antifungal activity against opportunistic pathogenic Candida species and Geotrichum capitatum. The treatment with this anthraquinone reduced hyphal growth, inhibited biofilm formation, and damaged mature Candida albicans biofilms. Real-time RT-PCR revealed that quinalizarin downregulated the expression of hyphae-related and biofilm-specific genes. The flow cytometry method used in the study showed that both apoptosis and necrosis were the physiological mechanisms of quinalizarin-induced C. albicans cell death, depending on the dose of the antifungal agent. A further study revealed an increase in the levels of intracellular reactive oxygen species and alterations in mitochondrial membrane potential after treatment with quinalizarin. Finally, quinalizarin was found to have low toxicity in a hemolytic test using human erythrocytes. In conclusion, we have identified quinalizarin as a potential antifungal compound.IMPORTANCEThis article is a study to determine the antifungal activity of quinalizarin (1,2,5,8-tetrahydroxyanthraquinone). Quinalizarin has potential antitumor properties and is effective in different types of tumor cells. The aim of the present study was to prove that quinalizarin can be used simultaneously in the treatment of cancer and in the treatment of intercurrent fungal infections. Quinalizarin was identified as a novel antifungal compound with low toxicity. These results may contribute to the development of a new drug with dual activity in the treatment of cancer-associated candidiasis.
Collapse
Affiliation(s)
- Monika Janeczko
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Elżbieta Kochanowicz
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Kamila Górka
- Department of Molecular Biology, Faculty of Medicine, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Tomasz Skrzypek
- Department of Biomedicine and Environmental Research, Faculty of Medicine, The John Paul II Catholic University of Lublin, Lublin, Poland
| |
Collapse
|
21
|
Liu Y, Zhang J, Chen H, Zhang W, Ainiwaer A, Mao S, Yao X, Xu T, Yan Y. Urinary microbiota signatures associated with different types of urinary diversion: a comparative study. Front Cell Infect Microbiol 2024; 13:1302870. [PMID: 38235491 PMCID: PMC10791864 DOI: 10.3389/fcimb.2023.1302870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/17/2023] [Indexed: 01/19/2024] Open
Abstract
Background Radical cystectomy and urinary diversion (UD) are gold standards for non-metastatic muscle-invasive bladder cancer. Orthotopic neobladder (or Studer), ileal conduit (or Bricker) and cutaneous ureterostomy (CU) are mainstream UD types. Little is known about urinary microbiological changes after UD. Methods In this study, urine samples were collected from healthy volunteers and patients with bladder cancer who had received aforementioned UD procedures. Microbiomes of samples were analyzed using 16S ribosomal RNA gene sequencing, and microbial diversities, distributions and functions were investigated and compared across groups. Results Highest urine microbial richness and diversity were observed in healthy controls, followed by Studer patients, especially those without hydronephrosis or residual urine, α-diversity indices of whom were remarkably higher than those of Bricker and CU groups. Studer UD type was the only independent factor favoring urine microbial diversity. The urine microflora structure of the Studer group was most similar to that of the healthy individuals while that of the CU group was least similar. Studer patients and healthy volunteers shared many similar urine microbial functions, while Bricker and CU groups exhibited opposite characteristics. Conclusion Our study first presented urinary microbial landscapes of UD patients and demonstrated the microbiological advantage of orthotopic neobladder. Microbiota might be a potential tool for optimization of UD management.
Collapse
Affiliation(s)
- Yuchao Liu
- Department of Urology, Chongming Branch of Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Jingcheng Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Haotian Chen
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Ailiyaer Ainiwaer
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Department of Urology, Kashgar Prefecture Second People Hospital, Kashgar, Xinjiang Uygur Autonomous Region, China
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Xudong Yao
- Department of Urology, Chongming Branch of Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Tianyuan Xu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| | - Yang Yan
- Department of Urology, Chongming Branch of Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Institute of Urinary Oncology, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
22
|
Tavakolian S, Tabaeian SP, Namazi A, Faghihloo E, Akbari A. Role of the VEGF in virus-associated cancers. Rev Med Virol 2024; 34:e2493. [PMID: 38078693 DOI: 10.1002/rmv.2493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/14/2023] [Indexed: 01/30/2024]
Abstract
The role of numerous risk factors, including consumption of alcohol, smoking, having diet high in fat and sugar and many other items, on caner progression cannot be denied. Viral diseases are one these factors, and they can initiate some signalling pathways causing cancer. For example, they can be effective on providing oxygen and nutrients by inducing VEGF expression. In this review article, we summarised the mechanisms of angiogenesis and VEGF expression in cancerous tissues which are infected with oncoviruses (Epstein-Barr virus, Human papillomavirus infection, Human T-lymphotropic virus, Kaposi's sarcoma-associated herpesvirus, Hepatitis B and hepatitis C virus).
Collapse
Affiliation(s)
- Shaian Tavakolian
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Seidamir Pasha Tabaeian
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Namazi
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Faghihloo
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
- Occupational Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Tognarelli EI, Gutiérrez-Vera C, Palacios PA, Pasten-Ferrada IA, Aguirre-Muñoz F, Cornejo DA, González PA, Carreño LJ. Natural Killer T Cell Diversity and Immunotherapy. Cancers (Basel) 2023; 15:5737. [PMID: 38136283 PMCID: PMC10742272 DOI: 10.3390/cancers15245737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Invariant natural killer T cells (iNKTs), a type of unconventional T cells, share features with NK cells and have an invariant T cell receptor (TCR), which recognizes lipid antigens loaded on CD1d molecules, a major histocompatibility complex class I (MHC-I)-like protein. This interaction produces the secretion of a wide array of cytokines by these cells, including interferon gamma (IFN-γ) and interleukin 4 (IL-4), allowing iNKTs to link innate with adaptive responses. Interestingly, molecules that bind CD1d have been identified that enable the modulation of these cells, highlighting their potential pro-inflammatory and immunosuppressive capacities, as required in different clinical settings. In this review, we summarize key features of iNKTs and current understandings of modulatory α-galactosylceramide (α-GalCer) variants, a model iNKT cell activator that can shift the outcome of adaptive immune responses. Furthermore, we discuss advances in the development of strategies that modulate these cells to target pathologies that are considerable healthcare burdens. Finally, we recapitulate findings supporting a role for iNKTs in infectious diseases and tumor immunotherapy.
Collapse
Affiliation(s)
- Eduardo I. Tognarelli
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Ignacio A. Pasten-Ferrada
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Fernanda Aguirre-Muñoz
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Daniel A. Cornejo
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Santiago 8330025, Chile; (E.I.T.); (C.G.-V.); (P.A.P.); (I.A.P.-F.); (F.A.-M.); (D.A.C.)
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| |
Collapse
|
24
|
Mansour O, Kazem A, El Wakil A. Assessment of breast cytoarchitecture and its associated axillary lymph node status under normal and pathological conditions in Egyptian women. Tissue Cell 2023; 85:102244. [PMID: 37856936 DOI: 10.1016/j.tice.2023.102244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 10/21/2023]
Abstract
OBJECTIVE Herein, we compare the features of neoplastic cancer cells in invasive ductal carcinoma (IDC) grade II and III patients to their corresponding normal cells both in breast and axillary lymph node (ALN) tissues. METHODS A retrospective cohort of 70 female breast cancer patients enrolled between 2018 and 2020 at Medical Research Institute, Alexandria University, Egypt, was analyzed for clinicopathological features presentation. Fresh tiny pieces of breast tissue and its associated ALN tissues were then processed to investigate the morphological appearance by scanning electron microscopy. Moreover, the histological architecture of tissue sections stained with hematoxylin and eosin was studied by light microscope, while the characterization of the ultrastructure features of breast and ALN tissues was analyzed by transmission electron microscopy. RESULTS Clinicopathological presentation of patients revealed that the Egyptian female breast cancer population adhered to the global trends of breast cancer disease with elevated incidence rate among postmenopausal women (61.3%), high frequency of IDC (95.7%), and increased ALN metastasis (65.7%). The percentage of estrogen receptor alpha (ERα) and human epidermal growth factor receptor 2 (HER2) expression, as key indicators for carcinogenesis and disease progression was 87.1% and 55.8%, respectively. The present study points to the observed discrepancies among the investigated variables in the diagnostic separation between IDC grade II and grade III. Ductal epithelial cells organization, nuclei size and irregularity, chromatin amount and uniformity, mitochondrial abundance and dysfunction were differentially manifested in IDC grades. Moreover, aberrations in the cellular organelles like lysosomes, endoplasmic reticulum, and lipid droplets vary according to the grade of IDC and the aggressiveness of the invasive breast cancer. CONCLUSIONS To sum up, this study emphasizes the importance of accurate specimen evaluation for treatment choice and decision.
Collapse
Affiliation(s)
- Omnia Mansour
- Department of Biological and Geological Sciences, Faculty of Education, Alexandria University, Egypt
| | - Amani Kazem
- Department of Pathology, Medical Research Institute, Alexandria University, Egypt
| | - Abeer El Wakil
- Department of Biological and Geological Sciences, Faculty of Education, Alexandria University, Egypt.
| |
Collapse
|
25
|
Aldosari BN, Abd El-Aal M, Abo Zeid EF, Faris TM, Aboelela A, Abdellatif AAH, Tawfeek HM. Synthesis and characterization of magnetic Ag-Fe 3O 4@polymer hybrid nanocomposite systems with promising antibacterial application. Drug Dev Ind Pharm 2023; 49:723-733. [PMID: 37906615 DOI: 10.1080/03639045.2023.2277812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION Bacterial infections caused by different strains of bacteria still one of the most important disorders affecting humans worldwide. Polymers nanocomposite systems could be considered as an alternative to conventional antibiotics to eradicate bacterial infections. SIGNIFICANCE In an attempt to enhance the antibacterial performance of silver and iron oxide nanoparticles, decrease their aggregation and toxicity, a polymeric hybrid nanocomposite system combining both nanoparticles is produced. METHODS Magnetic Ag-Fe3O4@polymer hybrid nanocomposites prepared using different polymers, namely polyethylene glycol 4000, ethyl cellulose, and chitosan were synthesized via wet impregnation and ball-milling techniques. The produced nanocomposites were tested for their physical properties and antibacterial activities. RESULTS XRD, FT-IR, VSM, and TEM results confirmed the successful preparation of hybrid nanocomposites. Hybrid nanocomposites have average crystallite sizes in the following order Ag-Fe3O4@CS (8.9 nm) < Ag-Fe3O4@EC (9.0 nm) < Ag-Fe3O4@PEG4000 (9.4 nm) and active surface area of this trend Ag-Fe3O4@CS (130.4 m2g-1) > Ag-Fe3O4@EC (128.9 m2g-1) > Ag-Fe3O4@PEG4000 (123.4 m2g-1). In addition, they have a saturation magnetization in this order: Ag-Fe3O4@PEG4000 (44.82 emu/g) > Ag-Fe3O4@EC (40.14 emu/g) > Ag-Fe3O4@CS (22.90 emu/g). Hybrid nanocomposites have a pronounced antibacterial action against Bacillus cereus, Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus intermedius compared to iron oxide nanoparticles and positive antibacterial drug. In addition, both Ag-Fe3O4@EC and Ag-Fe3O4@CS have a lower MIC values compared to Ag-Fe3O4@PEG and positive control. CONCLUSION Magnetic Ag-Fe3O4 hybrid nanocomposites could be promising antibacterial nanomaterials and could pave the way for the development of new materials with even more unique properties and applications.
Collapse
Affiliation(s)
- Basmah N Aldosari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed Abd El-Aal
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Essam F Abo Zeid
- Physics Department, Faculty of Science, Assiut University, Assiut, Egypt
- Biophysics Department, Faculty of Oral and Dental, Sphinx University, Assiut, Egypt
| | - Tarek M Faris
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ashraf Aboelela
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sphinx University, Assiut, Egypt
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Hesham M Tawfeek
- Industrial Pharmacy Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
26
|
Liu C, Zhao X, Wang Z, Zhao Y, Li R, Chen X, Chen H, Wan M, Wang X. Metal-organic framework-modulated Fe 3O 4 composite au nanoparticles for antibacterial wound healing via synergistic peroxidase-like nanozymatic catalysis. J Nanobiotechnology 2023; 21:427. [PMID: 37968680 PMCID: PMC10647143 DOI: 10.1186/s12951-023-02186-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/02/2023] [Indexed: 11/17/2023] Open
Abstract
Bacterial wound infections are a serious threat due to the emergence of antibiotic resistance. Herein, we report an innovative hybrid nanozyme independent of antibiotics for antimicrobial wound healing. The hybrid nanozymes are fabricated from ultra-small Au NPs via in-situ growth on metal-organic framework (MOF)-stabilised Fe3O4 NPs (Fe3O4@MOF@Au NPs, FMA NPs). The fabricated hybrid nanozymes displayed synergistic peroxidase (POD)-like activities. It showed a remarkable level of hydroxyl radicals (·OH) in the presence of a low dose of H2O2 (0.97 mM). Further, the hybrid FMA nanozymes exhibited excellent biocompatibility and favourable antibacterial effects against both Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria. The animal experiments indicated that the hybrid nanozymes promoted wound repair with adequate biosafety. Thus, the well-designed hybrid nanozymes represent a potential strategy for healing bacterial wound infections, without any toxic side effects, suggesting possible applications in antimicrobial therapy.
Collapse
Affiliation(s)
- Chuan Liu
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Xuanping Zhao
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Zichao Wang
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Yingyuan Zhao
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Ruifang Li
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Xuyang Chen
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Hong Chen
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Mengna Wan
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China
| | - Xueqin Wang
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, Henan, China.
- Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
27
|
Ding YY, Zhou H, Peng-Deng, Zhang BQ, Zhang ZJ, Wang GH, Zhang SY, Wu ZR, Wang YR, Liu YQ. Antimicrobial activity of natural and semi-synthetic carbazole alkaloids. Eur J Med Chem 2023; 259:115627. [PMID: 37467619 DOI: 10.1016/j.ejmech.2023.115627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/22/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023]
Abstract
Since the first natural carbazole alkaloid, murrayanine, was isolated from Mwraya Spreng, carbazole alkaloid derivatives have been widely concerned for their anti-tumor, anti-viral and anti-bacterial activities. In recent decades, a growing body of data suggest that carbazole alkaloids and their derivatives have different biological activities. This is the first comprehensive description of the antifungal and antibacterial activities of carbazole alkaloids in the past decade (2012-2022), including natural and partially synthesized carbazole alkaloids in the past decade. Finally, the challenges and problems faced by this kind of alkaloids are summarized. This paper will be helpful for further exploration of this kind of alkaloids.
Collapse
Affiliation(s)
- Yan-Yan Ding
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China; Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, 313000, China
| | - Han Zhou
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Peng-Deng
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Bao-Qi Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Zhi-Jun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Guang-Han Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Shao-Yong Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, 313000, China
| | - Zheng-Rong Wu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Yi-Rong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Ying-Qian Liu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China; Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, 313000, China; State Key Laboratory of Grassland Agro-ecosystems, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
28
|
Rossato Viana A, Nicola I, Franco C, Caetano PA, Jacob-Lopes E, Zepka LQ, Santos D, Moraes Flores EM, Stefanello Vizzotto B, Wolf K, Ferreira Ourique A, Mortari SR, Bohn Rhoden CR, Fontanari Krause LM. Phytochemical characterization and toxicological activity attributed to the acetonic extract of South American Vassobia breviflora. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2023; 86:816-832. [PMID: 37667472 DOI: 10.1080/15287394.2023.2254316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The particular plant species found in southern Brazil, Vassobia breviflora (Solanaceae) has only a few apparent studies examining its biological effect. Thus, the aim of the present study was to determine the activity of the acetone extract fraction derived from V. breviflora. Four compounds were identified by ESI-qTOF-MS: eucalrobusone R, aplanoic acid B, pheophorbide A, and pheophytin A. In addition, 5 compounds were identified by HPLC-PDA-MS/MS: all-trans-lutein, 15-cis-lutein, all-trans-β-carotene, 5,8-epoxy-β-carotene, and cis-β-carotene. Cell lines A549 (lung cancer), A375 (melanoma cancer) and HeLa (cervical cancer) were incubated with different concentrations of each studied extract using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), lactate dehydrogenase (LDH), and 2'-7'dichlorofluorescin diacetate (DCFH-DA) assays. The acetonic extract exhibited cytotoxic activity at a concentration of 0.03 mg/ml in the HeLa strain and 0.1 mg/ml in the others. In addition to increased production of reactive oxygen species (ROS). Antibacterial activity was assessed utilizing minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) in 9 ATCCs strains and 7 clinical isolates, as well as determination of biofilm production. Data demonstrated that MIC and MBC were approximately 256 mg/ml in most of the strains tested and antibiofilm effect at S. aureus, S. epidermidis, A. baumannii, and E. faecalis, concentrations below the MIC. Genotoxic activity on plasmid DNA did not produce significant elevated levels in breaks in the isolated genetic material.
Collapse
Affiliation(s)
- Altevir Rossato Viana
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Brazil
| | | | - Camila Franco
- Biomedicine, Franciscan University, Santa Maria, Brazil
| | - Patrícia Acosta Caetano
- Department of Food Science and Technology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Eduardo Jacob-Lopes
- Department of Food Science and Technology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Leila Queiroz Zepka
- Department of Food Science and Technology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Daniel Santos
- Department of Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | | | | | - Katianne Wolf
- Laboratory of Nanotechnology, Franciscan University, Santa Maria, Brazil
| | | | | | - Cristiano Rodrigo Bohn Rhoden
- Laboratory of Nanotechnology, Franciscan University, Santa Maria, Brazil
- Laboratory of Nanoesctructurated Magnetic Materials - LaMMaN, Nanosciences Post-graduation Program, Franciscan University, Santa Maria, Brazil
| | | |
Collapse
|
29
|
Choudhury A, Ortiz PS, Young M, Mahmud MT, Stoffel RT, Greathouse KL, Kearney CM. Control of Helicobacter pylori with engineered probiotics secreting selective guided antimicrobial peptides. Microbiol Spectr 2023; 11:e0201423. [PMID: 37712669 PMCID: PMC10580918 DOI: 10.1128/spectrum.02014-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/26/2023] [Indexed: 09/16/2023] Open
Abstract
Helicobacter pylori is the primary cause of 78% of gastric cancer cases, providing an opportunity to prevent cancer by controlling a single bacterial pathogen within the complex gastric microbiota. We developed highly selective antimicrobial agents against H. pylori by fusing an H. pylori-binding guide peptide (MM1) to broad-spectrum antimicrobial peptides. The common dairy probiotic Lactococcus lactis was then engineered to secrete these guided antimicrobial peptides (gAMPs). When co-cultured in vitro with H. pylori, the gAMP probiotics lost no toxicity compared to unguided AMP probiotics against the target, H. pylori, while losing >90% of their toxicity against two tested off-target bacteria. To test binding to H. pylori, the MM1 guide was fused to green fluorescent protein (GFP), resulting in enhanced binding compared to unguided GFP as measured by flow cytometry. In contrast, MM1-GFP showed no increased binding over GFP against five different off-target bacteria. These highly selective gAMP probiotics were then tested by oral gavage in mice infected with H. pylori. As a therapy, the probiotics outperformed antibiotic treatment, effectively eliminating H. pylori in just 5 days, and also protected mice from challenge infection as a prophylactic. As expected, the gAMP probiotics were as toxic against H. pylori as the unguided AMP probiotics. However, a strong rebound in gastric species diversity was found with both the selective gAMP probiotics and the non-selective AMP probiotics. Eliminating the extreme microbial dysbiosis caused by H. pylori appeared to be the major factor in diversity recovery. IMPORTANCE Alternatives to antibiotics in the control of Helicobacter pylori and the prevention of gastric cancer are needed. The high prevalence of H. pylori in the human population, the induction of microbial dysbiosis by antibiotics, and increasing antibiotic resistance call for a more sustainable approach. By selectively eliminating the pathogen and retaining the commensal community, H. pylori control may be achieved without adverse health outcomes. Antibiotics are typically used as a therapeutic post-infection, but a more targeted, less disruptive approach could be used as a long-term prophylactic against H. pylori or, by extension, against other gastrointestinal pathogens. Furthermore, the modular nature of the guided antimicrobial peptide (gAMP) technology allows for the substitution of different guides for different pathogens and the use of a cocktail of gAMPs to avoid the development of pathogen resistance.
Collapse
Affiliation(s)
| | | | - Mikaeel Young
- Department of Biology, Baylor University, Waco, Texas, USA
| | | | - Ryan T. Stoffel
- Baylor Sciences Building Vivarium, Baylor University, Waco, Texas, USA
| | - K. Leigh Greathouse
- Department of Biology, Baylor University, Waco, Texas, USA
- Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | | |
Collapse
|
30
|
Kılıç GA, Alsafi M. β-Glucan Regulates Lipopolysaccharide Induced Genotoxic Damage to The Liver through The Induction of BRCA1 Protein Expression. CELL JOURNAL 2023; 25:645-654. [PMID: 37718767 PMCID: PMC10520986 DOI: 10.22074/cellj.2023.1989382.1226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/20/2023] [Accepted: 05/14/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVE The present study aims to investigate the role of breast cancer-susceptibility gene 1 (BRCA1) protein in the β-Glucan (βG) molecule mediated regulation of lipopolysaccharide (LPS)-induced liver genotoxicity. MATERIALS AND METHODS In this experimental study, totally, 32 male Swiss Albino mice were randomly divided into 4 equal groups: control (C), LPS-administered (LPS), βG-administered (βG) and βG-pre-administered/LPS-administered (βG+LPS). The βG was injected at the dose of 150 mg/kg/day intraperitoneally (i.p.) for 3 days. A single dose of 4 mg/ kg (i.p.) LPS was administered 24 hours after the last βG injection. BRCA1 expression was determined by western blot analysis and confirmed by quantitative immunofluorescence. Proliferating cell nuclear antigen (PCNA), nuclear factor erythroid 2-related factor (Nrf2) and 8-OHdG protein levels were also determined by the immunofluorescence analysis. The alkaline comet assay was performed. superoxide dismutase (SOD), catalase (CAT) and membrane lipid peroxidation were biochemically measured, and light microscopic histology was evaluated. RESULTS The BRCA1 expression level was significantly decreased in the LPS group. However, in the βG+LPS group, expression of BRCA1 protein was over 2 folds higher than the control. After the LPS induction, the DNA strand breaks, oxidative DNA lesions and abnormal proliferation of the liver cells were almost entirely suppressed in βG preadministrated animals, indicating the BRCA1 mediated ubiquitination of PCNA and activation of the DNA damage repair pathways. Activation of Nrf2 in the βG+LPS group resulted in an increase in the levels of Nrf2 pathway dependent antioxidant enzymes SOD and CAT, prevented the peroxidation of membrane lipids and maintained the histological architecture of the liver. CONCLUSION The results manifested that the βG is a strong inducer of the BRCA1 protein expression in the LPSinduced hepatic stress and the protein constitutes the key component of a βG mediated liver protection against an LPS-induced genotoxic and pathological damage.
Collapse
Affiliation(s)
- Gözde Aydoğan Kılıç
- Department of Biology, Faculty of Science, Eskişehir Technical University, Eskişehir, Turkey
| | - Mojahed Alsafi
- Department of Biology, Faculty of Science, Eskişehir Technical University, Eskişehir, Turkey
| |
Collapse
|
31
|
Tavassoli M, Khezerlou A, Khalilzadeh B, Ehsani A, Kazemian H. Aptamer-modified metal organic frameworks for measurement of food contaminants: a review. Mikrochim Acta 2023; 190:371. [PMID: 37646854 DOI: 10.1007/s00604-023-05937-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023]
Abstract
The measurement of food contaminants faces a great challenge owing to the increasing demand for safe food, increasing consumption of fast food, and rapidly changing patterns of human consumption. As different types of contaminants in food products can pose different levels of threat to human health, it is desirable to develop specific and rapid methods for their identification and quantification. During the past few years, metal-organic framework (MOF)-based materials have been extensively explored in the development of food safety sensors. MOFs are porous crystalline materials with tunable composition, dynamic porosity, and facile surface functionalization. The construction of high-performance biosensors for a range of applications (e.g., food safety, environmental monitoring, and biochemical diagnostics) can thus be promoted through the synergistic combination of MOFs with aptamers. Accordingly, this review article delineates recent innovations achieved for the aptamer-functionalized MOFs toward the detection of food contaminants. First, we describe the basic concepts involved in the detection of food contaminants in terms of the advantages and disadvantages of the commonly used analytical methods (e.g., DNA-based methods (PCR/real-time PCR/multiplex PCR/digital PCR) and protein-based methods (enzyme-linked immunosorbent assay/immunochromatography assay/immunosensor/mass spectrometry). Afterward, the progress in aptamer-functionalized MOF biosensors is discussed with respect to the sensing mechanisms (e.g., the role of MOFs as signal probes and carriers for loading signal probes) along with their performance evaluation (e.g., in terms of sensitivity). We finally discuss challenges and opportunities associated with the development of aptamer-functionalized MOFs for the measurement of food contaminants.
Collapse
Affiliation(s)
- Milad Tavassoli
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Food Science and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezou Khezerlou
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Food Science and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Balal Khalilzadeh
- Stem Cell Research Center (SCRC), Tabriz University of Medical Sciences, Tabriz, 51666-14711, Iran
| | - Ali Ehsani
- Department of Food Science and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hossein Kazemian
- Materials Technology & Environmental Research (MATTER) Lab, University of Northern British Columbia, Prince George, BC, Canada.
- Northern Analytical Lab Services (Northern BC's Environmental and Climate Solutions Innovation Hub), University of Northern British Columbia, Prince George, BC, Canada.
- Environmental Sciences Program, Faculty of Environment, University of Northern British Columbia, Prince George, BC, V2N4Z9, Canada.
| |
Collapse
|
32
|
Ma T, Tang Y, Wang T, Yang Y, Zhang Y, Wang R, Zhang Y, Li Y, Wu M, Tang M, Hu X, Zou C, Ren Y, Liu H, Zhang Q, Li H, Wu M, Li J, Zhou X. Chronic pulmonary bacterial infection facilitates breast cancer lung metastasis by recruiting tumor-promoting MHCII hi neutrophils. Signal Transduct Target Ther 2023; 8:296. [PMID: 37563136 PMCID: PMC10415306 DOI: 10.1038/s41392-023-01542-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 08/12/2023] Open
Abstract
Breast cancer can metastasize to various organs, including the lungs. The immune microenvironment of the organs to be metastasized plays a crucial role in the metastasis of breast cancer. Infection with pathogens such as viruses and bacteria can alter the immune status of the lung. However, the effect of chronic inflammation caused by bacteria on the formation of a premetastatic niche within the lung is unclear, and the contribution of specific immune mediators to tumor metastasis also remains largely undetermined. Here, we used a mouse model revealing that chronic pulmonary bacterial infection augmented breast cancer lung metastasis by recruiting a distinct subtype of tumor-infiltrating MHCIIhi neutrophils into the lung, which exhibit cancer-promoting properties. Functionally, MHCIIhi neutrophils enhanced the lung metastasis of breast cancer in a cell-intrinsic manner. Furthermore, we identified CCL2 from lung tissues as an important environmental signal to recruit and maintain MHCIIhi neutrophils. Our findings clearly link bacterial-immune crosstalk to breast cancer lung metastasis and define MHCIIhi neutrophils as the principal mediator between chronic infection and tumor metastasis.
Collapse
Affiliation(s)
- Teng Ma
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yu Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Taolin Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yang Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yige Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Ruihuan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yongxin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yi Li
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, China
| | - Mingbo Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Miao Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Xueli Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Chaoyu Zou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yuan Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Qianhua Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Heyue Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Min Wu
- Drug Discovery Center, Wenzhou Institute, University of Chinese Academy of Sciences, 325001, Wenzhou, China.
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Xikun Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
33
|
Tustumi F, Arienzo VP, Sunye IR, Lucas PFS, Colonno BB, Quintas JG, Lisboa EN, Szor DJ. Esophageal Dysbiosis in Achalasia and Cancer Development: A Critical Review. Genes (Basel) 2023; 14:1521. [PMID: 37628573 PMCID: PMC10454429 DOI: 10.3390/genes14081521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Microorganisms provide various benefits to their human hosts, including assisting with digestion, synthesizing certain vitamins, developing the gastrointestinal and immune systems, regulating metabolism, and protecting against some pathogens. However, microbial imbalances can cause tissue damage and contribute to inflammatory disorders and cancers. Microbial dysbiosis refers to an imbalance or disruption in the normal composition and function of the microbial communities that inhabit various body parts, including the gut, oral cavity, skin, and reproductive tract. Emerging research suggests that microbial dysbiosis plays a significant role in cancer development and progression. This issue is particularly relevant in achalasia, in which food stasis, changes in endoluminal pH, and poor esophageal clearance might contribute to esophageal microbial dysbiosis. This study aimed to evaluate the association between dysbiosis and esophageal cancer development, focused on esophageal dysmotility disorders. METHODS This study is a critical review, gathering the current evidence for the association between dysbiosis and the development of esophageal cancer. RESULTS Studies have shown that microbiota play a role in cancer development, although the mechanisms for how they do so are not yet fully understood. One possible explanation is that microbiota alterations can lead to chronic inflammation, promoting cancer cell growth. Additionally, some bacteria produce toxins that can damage DNA and cause genomic instability, and certain bacterial products can promote tumor growth. CONCLUSION Despite the close relationship between dysbiosis and cancer development in esophageal dysmotility disorders, further investigations are still needed to elucidate the precise mechanisms by which dysbiosis contributes to cancer development and to identify potential therapeutic interventions targeting the microbiota to prevent or treat cancer.
Collapse
Affiliation(s)
- Francisco Tustumi
- Department of Health Sciences, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil (J.G.Q.)
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kato I. Bacterial, Viral and Parasitic Pathogens and Colorectal Cancer. Cancers (Basel) 2023; 15:3353. [PMID: 37444463 DOI: 10.3390/cancers15133353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Several viral, bacterial, and parasitic pathogens have been designated as human carcinogens by the World Health Organization [...].
Collapse
Affiliation(s)
- Ikuko Kato
- Department of Oncology and Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
35
|
Pan J, You W, Lu X, Wang S, You Z, Sun Y. GSPHI: A novel deep learning model for predicting phage-host interactions via multiple biological information. Comput Struct Biotechnol J 2023; 21:3404-3413. [PMID: 37397626 PMCID: PMC10314231 DOI: 10.1016/j.csbj.2023.06.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/04/2023] Open
Abstract
Emerging evidence suggests that due to the misuse of antibiotics, bacteriophage (phage) therapy has been recognized as one of the most promising strategies for treating human diseases infected by antibiotic-resistant bacteria. Identification of phage-host interactions (PHIs) can help to explore the mechanisms of bacterial response to phages and provide new insights into effective therapeutic approaches. Compared to conventional wet-lab experiments, computational models for predicting PHIs can not only save time and cost, but also be more efficient and economical. In this study, we developed a deep learning predictive framework called GSPHI to identify potential phage and target bacterium pairs through DNA and protein sequence information. More specifically, GSPHI first initialized the node representations of phages and target bacterial hosts via a natural language processing algorithm. Then a graph embedding algorithm structural deep network embedding (SDNE) was utilized to extract local and global information from the interaction network, and finally, a deep neural network (DNN) was applied to accurately detect the interactions between phages and their bacterial hosts. In the drug-resistant bacteria dataset ESKAPE, GSPHI achieved a prediction accuracy of 86.65 % and AUC of 0.9208 under the 5-fold cross-validation technique, significantly better than other methods. In addition, case studies in Gram-positive and negative bacterial species demonstrated that GSPHI is competent in detecting potential Phage-host interactions. Taken together, these results indicate that GSPHI can provide reasonable candidate sensitive bacteria to phages for biological experiments. The webserver of the GSPHI predictor is freely available at http://120.77.11.78/GSPHI/.
Collapse
Affiliation(s)
- Jie Pan
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an 710069, China
| | - Wencai You
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an 710069, China
| | - Xiaoliang Lu
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an 710069, China
| | - Shiwei Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an 710069, China
| | - Zhuhong You
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710129, China
| | - Yanmei Sun
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an 710069, China
| |
Collapse
|
36
|
Li W, Zhou X, Yuan S, Wang L, Yu L, Sun J, Chen J, Xiao Q, Wan Z, Zheng JS, Zhang CX, Larsson SC, Farrington SM, Law P, Houlston RS, Tomlinson I, Ding KF, Dunlop MG, Theodoratou E, Li X. Exploring the Complex Relationship between Gut Microbiota and Risk of Colorectal Neoplasia Using Bidirectional Mendelian Randomization Analysis. Cancer Epidemiol Biomarkers Prev 2023; 32:809-817. [PMID: 37012201 PMCID: PMC10233354 DOI: 10.1158/1055-9965.epi-22-0724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/07/2022] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Human gut microbiome has complex relationships with the host, contributing to metabolism, immunity, and carcinogenesis. METHODS Summary-level data for gut microbiota and metabolites were obtained from MiBioGen, FINRISK and human metabolome consortia. Summary-level data for colorectal cancer were derived from a genome-wide association study meta-analysis. In forward Mendelian randomization (MR), we employed genetic instrumental variables (IV) for 24 gut microbiota taxa and six bacterial metabolites to examine their causal relationship with colorectal cancer. We also used a lenient threshold for nine apriori gut microbiota taxa as secondary analyses. In reverse MR, we explored association between genetic liability to colorectal neoplasia and abundance of microbiota studied above using 95, 19, and 7 IVs for colorectal cancer, adenoma, and polyps, respectively. RESULTS Forward MR did not find evidence indicating causal relationship between any of the gut microbiota taxa or six bacterial metabolites tested and colorectal cancer risk. However, reverse MR supported genetic liability to colorectal adenomas was causally related with increased abundance of two taxa: Gammaproteobacteria (β = 0.027, which represents a 0.027 increase in log-transformed relative abundance values of Gammaproteobacteria for per one-unit increase in log OR of adenoma risk; P = 7.06×10-8), Enterobacteriaceae (β = 0.023, P = 1.29×10-5). CONCLUSIONS We find genetic liability to colorectal neoplasia may be associated with abundance of certain microbiota taxa. It is more likely that subset of colorectal cancer genetic liability variants changes gut biology by influencing both gut microbiota and colorectal cancer risk. IMPACT This study highlights the need of future complementary studies to explore causal mechanisms linking both host genetic variation with gut microbiome and colorectal cancer susceptibility.
Collapse
Affiliation(s)
- Wanxin Li
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan Zhou
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuai Yuan
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lijuan Wang
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Yu
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Sun
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Chen
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Xiao
- Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Ju-Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Cai-Xia Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Susanna C. Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susan M. Farrington
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Cancer Research UK Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Ian Tomlinson
- Cancer Research UK Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Ke-Feng Ding
- Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Malcolm G. Dunlop
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Cancer Research UK Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Evropi Theodoratou
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| |
Collapse
|
37
|
Pani G. Fusobacterium & Co. at the Stem of Cancer: Microbe-Cancer Stem Cell Interactions in Colorectal Carcinogenesis. Cancers (Basel) 2023; 15:cancers15092583. [PMID: 37174049 PMCID: PMC10177588 DOI: 10.3390/cancers15092583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Adult stem cells lie at the crossroads of tissue repair, inflammation, and malignancy. Intestinal microbiota and microbe-host interactions are pivotal to maintaining gut homeostasis and response to injury, and participate in colorectal carcinogenesis. Yet, limited knowledge is available on whether and how bacteria directly crosstalk with intestinal stem cells (ISC), particularly cancerous stem-like cells (CR-CSC), as engines for colorectal cancer initiation, maintenance, and metastatic dissemination. Among several bacterial species alleged to initiate or promote colorectal cancer (CRC), the pathobiont Fusobacterium Nucleatum has recently drawn significant attention for its epidemiologic association and mechanistic linkage with the disease. We will therefore focus on current evidence for an F. nucleatum-CRCSC axis in tumor development, highlighting the commonalities and differences between F. nucleatum-associated colorectal carcinogenesis and gastric cancer driven by Helicobacter Pylori. We will explore the diverse facets of the bacteria-CSC interaction, analyzing the signals and pathways whereby bacteria either confer "stemness" properties to tumor cells or primarily target stem-like elements within the heterogeneous tumor cell populations. We will also discuss the extent to which CR-CSC cells are competent for innate immune responses and participate in establishing a tumor-promoting microenvironment. Finally, by capitalizing on the expanding knowledge of how the microbiota and ISC crosstalk in intestinal homeostasis and response to injury, we will speculate on the possibility that CRC arises as an aberrant repair response promoted by pathogenic bacteria upon direct stimulation of intestinal stem cells.
Collapse
Affiliation(s)
- Giovambattista Pani
- Department of Translational Medicine and Surgery, Section of General Pathology, Faculty of Medicine, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, L. go A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
38
|
Higashi DL, Krieger MC, Qin H, Zou Z, Palmer EA, Kreth J, Merritt J. Who is in the driver's seat? Parvimonas micra: An understudied pathobiont at the crossroads of dysbiotic disease and cancer. ENVIRONMENTAL MICROBIOLOGY REPORTS 2023. [PMID: 36999244 DOI: 10.1111/1758-2229.13153] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Recent advances in our understanding of microbiome composition at sites of inflammatory dysbiosis have triggered a substantial interest in a variety of historically understudied bacteria, especially among fastidious obligate anaerobes. A plethora of new evidence suggests that these microbes play outsized roles in establishing synergistic polymicrobial infections at many different sites in the human body. Parvimonas micra is a prime example of such an organism. Despite being almost completely uncharacterized at the genetic level, it is one of the few species commonly detected in abundance at multiple mucosal sites experiencing either chronic or acute inflammatory diseases, and more recently, it has been proposed as a discriminating biomarker for multiple types of malignancies. In the absence of disease, P. micra is commonly found in low abundance, typically residing within the oral cavity and gastrointestinal tract. P. micra exhibits the typical features of an inflammophilic organism, meaning its growth actually benefits from active inflammation and inflammatory tissue destruction. In this mini-review, we will describe our current understanding of this underappreciated but ubiquitous pathobiont, specifically focusing upon the role of P. micra in polymicrobial inflammatory dysbiosis and cancer as well as the key emerging questions regarding its pathobiology. Through this timely work, we highlight Parvimonas micra as a significant driver of disease and discuss its unique position at the crossroads of dysbiosis and cancer.
Collapse
Affiliation(s)
- Dustin L Higashi
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Madeline C Krieger
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Hua Qin
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Zhengzhong Zou
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Elizabeth A Palmer
- Department of Pediatric Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Jens Kreth
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Justin Merritt
- Department of Restorative Dentistry, Oregon Health and Science University, Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
39
|
Ding Q, Zhang W, Guo Y, Wang J, Chen H, Zhang L. β-Lactamase Sensitive Probe for Rapid Detection of Antibiotic-Resistant Bacteria with Gas Chromatography–Tandem Mass Spectrometry. Anal Chem 2023; 95:6098-6106. [PMID: 36972326 DOI: 10.1021/acs.analchem.3c00381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
β-Lactamase (Bla) produced by bacteria to resist β-lactam antibiotics is a serious public health threat. Developing efficient diagnostic protocols for drug-resistant bacteria is of great significance. In this work, based on gas molecules in bacteria, a novel research strategy was proposed to develop a gas molecule-based probe by grafting 2-methyl-3-mercaptofuran (MF) onto cephalosporin intermediates via a nucleophilic substitution reaction. The probe can release the corresponding MF by reacting with Bla. The released MF, as a marker of drug-resistant bacteria, was analyzed by headspace solid-phase microextraction coupled with gas chromatography-mass spectrometry. The Bla concentration as low as 0.2 nM can be easily observed, providing an efficient method for detecting enzyme activity and screening drug-resistant strains in vivo. Importantly, the method is universal, and probes with different properties can be prepared by changing different substrates to further identify different types of bacteria, thereby broadening the research methods and ideas for monitoring physiological processes.
Collapse
|
40
|
Mishra A, Maurya SK, Singh A, Siddique H, Samanta SK, Mishra N. Neolamarckia cadamba (Roxb.) Bosser (Rubiaceae) extracts: promising prospects for anticancer and antibacterial potential through in vitro and in silico studies. Med Oncol 2023; 40:99. [PMID: 36808013 DOI: 10.1007/s12032-023-01971-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/06/2023] [Indexed: 02/23/2023]
Abstract
Neolamarckia cadamba is an Indian traditional medicinal plant having various therapeutic potentials. In the present study, we did solvent-based extraction of Neolamarckia cadamba leaves. The extracted samples were screened against liver cancer cell line (HepG2) and bacteria (Escherichia coli). MTT cytotoxic assay was performed for in vitro analysis of extracted samples against the HepG2 cell lines and the normal human prostate PNT2 cell line. Chloroform extract of Neolamarckia cadamba leaves showed better activity with IC50 value 69 μg/ml. DH5α strain of Escherichia coli (E. coli) was cultured in Luria Bertani (LB) broth media and minimum inhibitory concentration (MIC) and Minimum bactericidal concentration (MBC) were calculated. Solvent extract chloroform showed better activity in MTT analysis and antibacterial screening and it was taken for characterization of phytocomposition by Fourier transform infrared (FTIR) and gas chromatography mass spectrometry (GC-MS). The identified phytoconstituents were docked with potential targets of liver cancer and E. coli. The phytochemical 1-(5-Hydroxy-6-hydroxymethyl-tetrahydropyran-2-yl)-5-methyl-1H-pyrimidine-2,4-dione shows highest docking score against the targets PDGFRA (PDB ID: 6JOL) and Beta-ketoacyl synthase 1(PDB ID: 1FJ4) and their stability was further confirmed by molecular dynamics simulation studies.
Collapse
Affiliation(s)
- Anamika Mishra
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Santosh Kumar Maurya
- Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Anirudh Singh
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Hifzur Siddique
- Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Sintu Kumar Samanta
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Nidhi Mishra
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India.
| |
Collapse
|
41
|
Gomez K, Schiavoni G, Nam Y, Reynier JB, Khamnei C, Aitken M, Palmieri G, Cossu A, Levine A, van Noesel C, Falini B, Pasqualucci L, Tiacci E, Rabadan R. Genomic landscape of virus-associated cancers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.14.23285775. [PMID: 36824731 PMCID: PMC9949223 DOI: 10.1101/2023.02.14.23285775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
It has been estimated that 15%-20% of human cancers are attributable to infections, mostly by carcinogenic viruses. The incidence varies worldwide, with a majority affecting developing countries. Here, we present a comparative analysis of virus-positive and virus-negative tumors in nine cancers linked to five viruses. We find that virus-positive tumors occur more frequently in males and show geographical disparities in incidence. Genomic analysis of 1,658 tumors reveals virus-positive tumors exhibit distinct mutation signatures and driver gene mutations and possess a lower somatic mutation burden compared to virus-negative tumors of the same cancer type. For example, compared to the respective virus-negative counterparts, virus-positive cases across different cancer histologies had less often mutations of TP53 and deletions of 9p21.3/ CDKN2 A- CDKN1A ; Epstein-Barr virus-positive (EBV+) gastric cancer had more frequent mutations of EIF4A1 and ARID1A and less marked mismatch repair deficiency signatures; and EBV-positive cHL had fewer somatic genetic lesions of JAK-STAT, NF-κB, PI3K-AKT and HLA-I genes and a less pronounced activity of the aberrant somatic hypermutation signature. In cHL, we also identify germline homozygosity in HLA class I as a potential risk factor for the development of EBV-positive Hodgkin lymphoma. Finally, an analysis of clinical trials of PD-(L)1 inhibitors in four virus-associated cancers suggested an association of viral infection with higher response rate in patients receiving such treatments, which was particularly evident in gastric cancer and head and neck squamous cell carcinoma. These results illustrate the epidemiological, genetic, prognostic, and therapeutic trends across virus-associated malignancies.
Collapse
|
42
|
Development Of Amoxicillin Trihydrate-Loaded Lyotropic Liquid Crystal Nanoparticles For Skin Infection. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
43
|
Watson MM, van der Giezen M, Søreide K. Gut Microbiome Influence on Human Epigenetics, Health, and Disease. HANDBOOK OF EPIGENETICS 2023:669-686. [DOI: 10.1016/b978-0-323-91909-8.00012-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
44
|
Investigational Microbiological Therapy for Glioma. Cancers (Basel) 2022; 14:cancers14235977. [PMID: 36497459 PMCID: PMC9736089 DOI: 10.3390/cancers14235977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022] Open
Abstract
Glioma is the most common primary malignancy of the central nervous system (CNS), and 50% of patients present with glioblastoma (GBM), which is the most aggressive type. Currently, the most popular therapies are progressive chemotherapy and treatment with temozolomide (TMZ), but the median survival of glioma patients is still low as a result of the emergence of drug resistance, so we urgently need to find new therapies. A growing number of studies have shown that the diversity, bioactivity, and manipulability of microorganisms make microbial therapy a promising approach for cancer treatment. However, the many studies on the research progress of microorganisms and their derivatives in the development and treatment of glioma are scattered, and nobody has yet provided a comprehensive summary of them. Therefore, in this paper, we review the research progress of microorganisms and their derivatives in the development and treatment of glioma and conclude that it is possible to treat glioma by exogenous microbial therapies and targeting the gut-brain axis. In this article, we discuss the prospects and pressing issues relating to these therapies with the aim of providing new ideas for the treatment of glioma.
Collapse
|
45
|
Saad MH, El-Moselhy TF, S El-Din N, Mehany ABM, Belal A, Abourehab MAS, Tawfik HO, El-Hamamsy MH. Discovery of new symmetrical and asymmetrical nitrile-containing 1,4-dihydropyridine derivatives as dual kinases and P-glycoprotein inhibitors: synthesis, in vitro assays, and in silico studies. J Enzyme Inhib Med Chem 2022; 37:2489-2511. [PMID: 36093880 PMCID: PMC9481151 DOI: 10.1080/14756366.2022.2120478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Two new series of symmetric (1a-h) and asymmetric (2a-l) 1,4-DHP derivatives were designed, synthesised, and evaluated as anticancer agents. In vitro anticancer screening of target compounds via National cancer institute “NCI” revealed that analogues 1g, 2e, and 2l demonstrated antiproliferative action with mean growth inhibition percentage “GI%” = 41, 28, and 64, respectively. The reversal doxorubicin (DOX) effects of compounds 1g, 2e, and 2l were examined and illustrated better cytotoxic activity with IC50 =1.12, 3.64, and 3.57 µM, respectively. The most active anticancer analogues, 1g, 2e, and 2l, were inspected for their putative mechanism of action by estimating their epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER-2), and Bruton’s tyrosine kinase (BTK) inhibitory activities. Furthermore, the antimicrobial activity of target compounds was assessed against six different pathogens, followed by determining the minimum inhibitory concentration “MIC” values for the most active analogues. Molecular docking study was achieved to understand mode of interactions between selected inhibitors and different biological targets.
Collapse
Affiliation(s)
| | - Tarek F El-Moselhy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Nabaweya S El-Din
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ahmed B M Mehany
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Amany Belal
- Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.,Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mervat H El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
46
|
Magneto-Fluorescent Mesoporous Nanocarriers for the Dual-Delivery of Ofloxacin and Doxorubicin to Tackle Opportunistic Bacterial Infections in Colorectal Cancer. Int J Mol Sci 2022; 23:ijms232012287. [PMID: 36293142 PMCID: PMC9603674 DOI: 10.3390/ijms232012287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/09/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer-related opportunistic bacterial infections are one major barrier for successful clinical therapies, often correlated to the production of genotoxic factors and higher cancer incidence. Although dual anticancer and antimicrobial therapies are a growing therapeutic fashion, they still fall short when it comes to specific delivery and local action in in vivo systems. Nanoparticles are seen as potential therapeutic vectors, be it by means of their intrinsic antibacterial properties and effective delivery capacity, or by means of their repeatedly reported modulation and maneuverability. Herein we report on the production of a biocompatible, antimicrobial magneto-fluorescent nanosystem (NANO3) for the delivery of a dual doxorubicin-ofloxacin formulation against cancer-related bacterial infections. The drug delivery capacity, rendered by its mesoporous silica matrix, is confirmed by the high loading capacity and stimuli-driven release of both drugs, with preference for tumor-like acidic media. The pH-dependent emission of its surface fluorescent SiQDs, provides an insight into NANO3 surface behavior and pore availability, with the SiQDs working as pore gates. Hyperthermia induces heat generation to febrile temperatures, doubling drug release. NANO3-loaded systems demonstrate significant antimicrobial activity, specifically after the application of hyperthermia conditions. NANO3 structure and antimicrobial properties confirm their potential use in a future dual anticancer and antimicrobial therapeutical vector, due to their drug loading capacity and their surface availability for further modification with bioactive, targeting species.
Collapse
|
47
|
Markelova NN, Semenova EF, Sineva ON, Sadykova VS. The Role of Cyclomodulins and Some Microbial Metabolites in Bacterial Microecology and Macroorganism Carcinogenesis. Int J Mol Sci 2022; 23:ijms231911706. [PMID: 36233008 PMCID: PMC9570213 DOI: 10.3390/ijms231911706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
A number of bacteria that colonize the human body produce toxins and effectors that cause changes in the eukaryotic cell cycle—cyclomodulins and low-molecular-weight compounds such as butyrate, lactic acid, and secondary bile acids. Cyclomodulins and metabolites are necessary for bacteria as adaptation factors—which are influenced by direct selection—to the ecological niches of the host. In the process of establishing two-way communication with the macroorganism, these compounds cause limited damage to the host, despite their ability to disrupt key processes in eukaryotic cells, which can lead to pathological changes. Possible negative consequences of cyclomodulin and metabolite actions include their potential role in carcinogenesis, in particular, with the ability to cause DNA damage, increase genome instability, and interfere with cancer-associated regulatory pathways. In this review, we aim to examine cyclomodulins and bacterial metabolites as important factors in bacterial survival and interaction with the host organism to show their heterogeneous effect on oncogenesis depending on the surrounding microenvironment, pathological conditions, and host genetic background.
Collapse
Affiliation(s)
- Natalia N. Markelova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| | - Elena F. Semenova
- Institute of Biochemical Technology, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, 295007 Simferopol, Russia
| | - Olga N. Sineva
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
| | - Vera S. Sadykova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| |
Collapse
|
48
|
Exosomal miRNA-21 from Toxoplasma gondii-infected microglial cells induces the growth of U87 glioma cells by inhibiting tumor suppressor genes. Sci Rep 2022; 12:16450. [PMID: 36180486 PMCID: PMC9525672 DOI: 10.1038/s41598-022-20281-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Toxoplasma gondii is an intracellular protozoan parasite that can modulate the microenvironment of infected hosts and is known to be associated with the incidence of brain tumor growth. In this study, we suggested that the exosomal microRNA-21 derived from Toxoplasma infection would contribute to the growth of brain tumors. Exosomes of BV2 microglial cells infected with Toxoplasma were characterized and confirmed internalization to U87 glioma cells. Exosomal miRNA expression profiles were analyzed using microRNA array and miR-21A-5p associated with Toxoplasma and tumor sorted. We also examined the mRNA level of tumor-associated genes in U87 glioma cells by changing the level of miR-21 within exosomes and the effects of exosomes on the proliferation of human U87 glioma cells. Expression of miRNA-21 was increased and anti-tumorigenic genes (FoxO1, PTEN, and PDCD4) were decreased in exosomes within T. gondii-infected U87 glioma cells. Toxoplasma-infected BV2-derived exosomes induced proliferation of U87 glioma cells. The exosomes induced the growth of U87 cells in a mouse tumor model. We suggest that the increased exosomal miR-21 from Toxoplasma-infected BV2 microglial cells may play an important role as a cell growth promotor of U87 glioma cells through a down-regulation of anti-tumorigenic genes.
Collapse
|
49
|
de Moraes LFRN, Silva PSE, Pereira TCPL, Almeida Rodrigues TA, Farias Frihling BE, da Costa RA, Torquato HFV, Lima CS, Paredes-Gamero EJ, Migliolo L. First generation of multifunctional peptides derived from latarcin-3a from Lachesana tarabaevi spider toxin. Front Microbiol 2022; 13:965621. [PMID: 36212827 PMCID: PMC9532841 DOI: 10.3389/fmicb.2022.965621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
The need for discovering new compounds that can act selectively on pathogens is becoming increasingly evident, given the number of deaths worldwide due to bacterial infections or tumor cells. New multifunctional biotechnological tools are being sought, including compounds present in spider venoms, which have high biotechnological potential. The present work aims to perform the rational design and functional evaluation of synthetic peptides derived from Lachesana tarabaevi spider toxin, known as latarcin-3a. The antimicrobial activity was tested against Gram-positive and -negative bacteria, with minimum inhibitory concentrations (MIC) between 4 and 128 μg.ml−1. Anti-biofilm tests were then performed to obtain MICs, where the peptides demonstrated activity from 4 to 128 μg.ml−1. In vitro cell cytotoxicity assays were carried out from tumor cell lines, lineages C1498, Kasumi-1, K-562, Jurkat, MOLT4, and Raji. Erythrocyte integrity was evaluated in the presence of synthetic peptides analog, which did not promote hemolysis at 128 μg.ml−1. The peptide that showed the best antibacterial activity was Lt-MAP3 and the best antitumor was Lt-MAP2. In conclusion, rational design of multifunctional antimicrobial peptides may be promising alternative tools in the treatment of emerging diseases such as bacterial infections and tumor cells.
Collapse
Affiliation(s)
- Luiz Filipe Ramalho Nunes de Moraes
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
- Selkis Biotech, Startup, Laboratório de Artrópodes Peçonhentos, Campo Grande, MS, Brazil
| | - Patrícia Souza e Silva
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | | | | | - Breno Emanuel Farias Frihling
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
- Selkis Biotech, Startup, Laboratório de Artrópodes Peçonhentos, Campo Grande, MS, Brazil
| | - Rosiane Andrade da Costa
- Programa de Pós-Graduação em Ciências Genômica e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | | | - Cauê Santos Lima
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Edgar Julian Paredes-Gamero
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Ludovico Migliolo
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
- Selkis Biotech, Startup, Laboratório de Artrópodes Peçonhentos, Campo Grande, MS, Brazil
- Programa de Pós-Graduação em Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
- *Correspondence: Ludovico Migliolo,
| |
Collapse
|
50
|
Ma R, Hu X, Zhang X, Wang W, Sun J, Su Z, Zhu C. Strategies to prevent, curb and eliminate biofilm formation based on the characteristics of various periods in one biofilm life cycle. Front Cell Infect Microbiol 2022; 12:1003033. [PMID: 36211965 PMCID: PMC9534288 DOI: 10.3389/fcimb.2022.1003033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Biofilms are colonies of bacteria embedded inside a complicated self-generating intercellular. The formation and scatter of a biofilm is an extremely complex and progressive process in constant cycles. Once formed, it can protect the inside bacteria to exist and reproduce under hostile conditions by establishing tolerance and resistance to antibiotics as well as immunological responses. In this article, we reviewed a series of innovative studies focused on inhibiting the development of biofilm and summarized a range of corresponding therapeutic methods for biological evolving stages of biofilm. Traditionally, there are four stages in the biofilm formation, while we systematize the therapeutic strategies into three main periods precisely:(i) period of preventing biofilm formation: interfering the colony effect, mass transport, chemical bonds and signaling pathway of plankton in the initial adhesion stage; (ii) period of curbing biofilm formation:targeting several pivotal molecules, for instance, polysaccharides, proteins, and extracellular DNA (eDNA) via polysaccharide hydrolases, proteases, and DNases respectively in the second stage before developing into irreversible biofilm; (iii) period of eliminating biofilm formation: applying novel multifunctional composite drugs or nanoparticle materials cooperated with ultrasonic (US), photodynamic, photothermal and even immune therapy, such as adaptive immune activated by stimulated dendritic cells (DCs), neutrophils and even immunological memory aroused by plasmocytes. The multitargeted or combinational therapies aim to prevent it from developing to the stage of maturation and dispersion and eliminate biofilms and planktonic bacteria simultaneously.
Collapse
Affiliation(s)
| | | | | | | | | | - Zheng Su
- *Correspondence: Chen Zhu, ; Zheng Su,
| | - Chen Zhu
- *Correspondence: Chen Zhu, ; Zheng Su,
| |
Collapse
|